Biosimilars: What Are They & Why Are They Important?

Ana Reis, PhD Ana Reis, PhD 7 min read

In healthcare, the introduction of biosimilars has become a significant pharmaceutical development that has made a tremendous impact, offering more affordable solutions to consumers. A biosimilar refers to a biologic medical product closely resembling an original product but manufactured by a distinct company. Biosimilars are officially sanctioned versions of the initial "innovator" products and are typically developed following the expiration of patents associated with the original product.

The era of these molecular “clones”, designed to closely mirror well-established biologic drugs, began in 2006 with the approval of Omnitrope, a recombinant human growth hormone, by the EMA in Europe, followed by the approval of Filgrastim-sndz (Zarxio) in the US in 2015, solidifying their significance across markets. Multiple drugs that are biologically similar increase the consumers options making drugs more affordable.

Understanding what biosimilars are, their role, and their importance can empower researchers, patients, healthcare professionals, and systems alike.

What Are Biosimilars in Medicine?

A biosimilar is a type of biologic medication that is highly similar to an already approved original biologic, known as the reference product or ‘innovator’ product. These biologic medications are made from the same types of sources (such as living cells or microorganisms) and exhibit no clinically meaningful differences from the reference product.

Many biosimilars are versions of monoclonal antibodies that are used to make biological drugs widely used in various therapeutic areas. Biosimilar antibodies are an important part of the domain, offering multifaceted applications in combating illnesses ranging from cancer to autoimmune disorders. Beyond their clinical utility, they also play a pivotal role in driving forward research and therapeutic interventions. 

Characteristics

Biologics, including biosimilars, are more complex than traditional drugs. Unlike generic drugs – chemical compounds that are identical replicas of their brand-name counterparts, biosimilars are complex molecules derived from living, natural sources, such as animal and plant cells or microorganisms. 

The purification, processing, and manufacturing of a biosimilar medicine is a complex, intricate process. Biosimilars undergo rigorous evaluation and testing to ensure they are highly similar to the original product (the reference product) in terms of quality, safety, and efficacy. 

Uses of Biosimilar Drugs: What Diseases Do Biosimilars Treat?

Biosimilars are used to treat various severe and chronic conditions, including:

  • Chronic skin diseases (such as psoriasis)
  • Chronic bowel diseases (such as Crohn’s disease and irritable bowel disorder)
  • Diabetes
  • Autoimmune conditions
  • Macular degeneration
  • Rheumatoid arthritis
  • Kidney conditions
  • Multiple sclerosis
  • Some cancers (such as breast, lung, and colon)

Biosimilars vs. Generic Medications: What’s the Difference?

While both biosimilars and generic medications aim to provide more affordable alternatives to their brand-name counterparts, there are differences between the two. Understanding these distinctions can help make informed choices.

  • Composition and Structure: Generic medications are chemically identical replicas of their reference drugs, typically small-molecule drugs with simple structures, and are produced through chemical synthesis. In contrast, biosimilars are highly similar but not identical to their reference biologic drugs, which are large, complex molecules derived from living systems. 
  • Regulation and Approval: Generic versions follow a shorter approval process called an Abbreviated New Drug Application. Biosimilars undergo a more extensive approval process compared to generic drugs. They are regulated differently and have significantly higher research and development costs and potential risks. The FDA’s Office of Generic Drugs reviews and approves new generic medicines, while biosimilars have their own approval pathway.

Why Are Biosimilar Drugs Important? Benefits of Biosimilars

From widening access to lifesaving treatments to fostering innovation in healthcare, biosimilars are not just alternatives but game-changers in the realm of medicine. Let’s break down the key advantages they bring to the table.

Lower-Cost Medicines for Affordable Healthcare

Similar to how generic drugs revolutionized the pharmaceutical industry, biosimilars introduce market competition. By providing alternatives to expensive biologics, they can drive down costs and enhance patient access to critical medicines, even for potentially deadly diseases.

The reduced cost to the consumer makes lifesaving medicines accessible to more people ensuring effective drugs are within reach of patients who need them. By providing lower-cost options, they contribute to more sustainable healthcare systems.

Increased Treatment Options

Biosimilars expand the range of treatment options available to patients, offering additional choices in their treatment plans. They allow healthcare providers to tailor treatments more effectively, considering both clinical efficacy and economic factors. This increased competition encourages innovation and can drive down costs for all biologic treatments.

Who Benefits from Biosimilars?

Biosimilars offer advantages to various stakeholders in the healthcare ecosystem:

  • Patients: Biosimilars are typically more affordable than reference biologics, and these cost savings can make critical treatments accessible to a broader population, especially when insurance coverage is limited, allowing patients to choose from multiple effective therapies.
  • Healthcare Systems and Payers: Biosimilars help manage healthcare budgets by reducing expenses associated with expensive biologics, and cost savings from biosimilars can be redirected to other healthcare needs.
  • Physicians and Healthcare Providers: Physicians can choose from a wider range of therapies, tailoring treatments to individual patient needs.
  • Biopharmaceutical Industry: Biosimilars encourage competition, driving innovation and improving overall drug development. Pharmaceutical companies can diversify their portfolios by developing both reference biologics and biosimilars.

Downsides

What are the challenges of biosimilars? While biosimilars offer cost-saving potential, as with any biologic medicine, there are certain drawbacks to address.

  • Complex Manufacturing Process: Biosimilars are created using living cells, making their manufacturing process more complex than that of traditional small-molecule generics. Ensuring consistency and similarity to the reference biologic can be challenging due to variations in cell lines, protein folding, and post-translational modifications.
  • Immunogenicity: Although biosimilars aim to be highly similar to the reference biologic, subtle differences can lead to immune responses. Some patients may develop antibodies against the biosimilar, affecting its efficacy or safety.
  • Market Share Challenges: Establishing a significant market share for biosimilars can be difficult. Physicians may prefer prescribing reference biologics, and patients may be hesitant to switch from a known biologic to a biosimilar.
  • Temperature Sensitivity: Biosimilars are sensitive to temperature variations. They often require distribution through a cold chain network to maintain stability and efficacy.
  • Lot-to-Lot Variability: Even among the same biosimilar product, there can be variations between different production lots which may impact side effects or efficacy.

Boost your yields! Get your free transient expression handbook to learn how!

Get it Now

The Manufacturing Process: How Are Biosimilars Produced?

The development of biosimilars as biotherapeutic products involves rigorous characterization, process optimization, confirmation of similarity, and regulatory approval to ensure safe and effective alternatives to reference biologics.

1. Characterization and Perfecting the Process

The initial step involves thoroughly understanding the reference biologic or product structure and function to identify critical quality attributes (CQAs).

2. Developing the Manufacturing

Using state-of-the-art biological development technologies and highly sensitive analytical tools, the biosimilar developer systematically engineers a molecule that closely matches the reference biologic’s quality attributes. This iterative process optimizes each part of the manufacturing procedure until the biosimilar consistently produces a highly similar molecular structure to the reference medicine.

3. Biosimilar Confirmation via Studies and Regulatory Cooperation

Once similarity has been established through analysis and testing, the next stage begins. Since biosimilars are prescription drugs, in the US, the U.S. Food and Drug Administration (FDA) reviews all information and determines additional non-clinical and clinical studies required to confirm biosimilarity and interchangeability.

4. Clinical Trials

Clinical trials are generally necessary for biosimilar approval in highly regulated markets (such as the EU, United States, Japan, Canada, and Australia). Clinical testing requirements and scope depend on the submitted data. If robust analytical data exist, tailored clinical trial programs may effectively demonstrate biosimilarity and interchangeability.

At ProteoGenix, our team of experts excels in every facet of antibody production, development, engineering, and characterization, catering to projects of all scales with our tailored custom antibody services.

The Approval Process for Biosimilar Medicinal Products

Let’s shed some light on the steps, regulations, and evaluations that govern the pathway of biosimilar drugs from inception to market.

  • Initial Approval: Biosimilars undergo a comprehensive approval process by regulatory agencies, such as the FDA in the United States or the EMA in Europe. The approval process involves demonstrating that the biosimilar is highly similar to the reference product with no clinically meaningful differences in terms of quality, safety, efficacy, or potency. Nevertheless, the FDA does not oversee whether health insurance companies include or compensate for the expenses associated with biosimilars in their health insurance plans.
  • Interchangeable Products: Some biosimilars may also receive an additional designation of interchangeability, meaning they can be substituted for the reference product without the intervention of the healthcare provider.
  • Ensuring Effectiveness After Approval: Post-approval surveillance ensures ongoing monitoring to confirm biosimilar drug safety and effectiveness in real-world settings. This continuous evaluation maintains confidence in biosimilars and ensures the reliability of finished products in clinical practice.

FDA-Approved Biosimilar Products

What biosimilars are on the market? Here are some FDA-approved biosimilar drugs along with their reference products:

  • Tyruko (natalizumab-sztn): Approved in August 2023, a biosimilar to Tysabri (natalizumab);
  • Yuflyma (adalimumab-aaty): Approved in May 2023, a biosimilar to Humira (adalimumab);
  • Idacio (adalimumab-aacf): Approved in December 2022, also a biosimilar to Humira (adalimumab);
  • Vegzelma (bevacizumab-adcd): Approved in September 2022, a biosimilar to Avastin (bevacizumab);
  • Stimufend (pegfilgrastim-fpgk): Approved in September 2022, a biosimilar to Neulasta (pegfilgrastim);
  • Cimerli (ranibizumab-eqrn): Approved in August 2022, a biosimilar to Lucentis (ranibizumab);
  • Fylnetra (pegfilgrastim-pbbk): Approved in May 2022, another biosimilar to Neulasta (pegfilgrastim).

Biosimilars as Catalysts for Affordable Healthcare

Biosimilars represent a promising frontier in healthcare, offering lower-cost and effective alternatives to original biological medicines. Through a rigorous approval process, biosimilars provide additional treatment options and contribute to more affordable healthcare.

As research and development continue, these medications will play an increasingly vital role in improving patient outcomes, affordability, and accessibility. Thus, everyone involved should stay informed about biosimilars’ potential impact on the medical landscape.

Need some for your project? Explore the diverse range of almost 2000 biosimilar therapeutic antibodies and over 70 biosimilar therapeutic proteins available at ProteoGenix – solutions that promise efficacy, safety, and affordability to meet your unique research needs.

Take the next step towards accessible and innovative treatment options. Feel free to get in touch to learn more – we’re here to serve as your trusted partner in achieving your goals.


Author Author: Ana Reis, PhD

Ana Reis is a scientific content manager at ProteoGenix. She has a PhD degree in biotechnology and a master's degree in science communication. Throughout her academic career, she has co-authored dozens of scientific articles on topics ranging from bioinformatics to chemical engineering.